EP2387571A1 - Oxadiazolderivate als slpl-rezeptoragonisten - Google Patents

Oxadiazolderivate als slpl-rezeptoragonisten

Info

Publication number
EP2387571A1
EP2387571A1 EP10700383A EP10700383A EP2387571A1 EP 2387571 A1 EP2387571 A1 EP 2387571A1 EP 10700383 A EP10700383 A EP 10700383A EP 10700383 A EP10700383 A EP 10700383A EP 2387571 A1 EP2387571 A1 EP 2387571A1
Authority
EP
European Patent Office
Prior art keywords
groups
group
tetrahydroisoquinolin
oxadiazol
dimethyl
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP10700383A
Other languages
English (en)
French (fr)
Inventor
Nuria Aguilar Izquierdo
Marta Carrascal Riera
Julio Cesar Castro Palomino Laria
Montserrat Erra Sola
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Almirall SA
Original Assignee
Almirall SA
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Almirall SA filed Critical Almirall SA
Priority to EP10700383A priority Critical patent/EP2387571A1/de
Publication of EP2387571A1 publication Critical patent/EP2387571A1/de
Withdrawn legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/02Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings
    • C07D413/04Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing two hetero rings directly linked by a ring-member-to-ring-member bond
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/435Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with one nitrogen as the only ring hetero atom
    • A61K31/47Quinolines; Isoquinolines
    • A61K31/472Non-condensed isoquinolines, e.g. papaverine
    • A61K31/4725Non-condensed isoquinolines, e.g. papaverine containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P11/00Drugs for disorders of the respiratory system
    • A61P11/06Antiasthmatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P17/00Drugs for dermatological disorders
    • A61P17/06Antipsoriatics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P19/00Drugs for skeletal disorders
    • A61P19/02Drugs for skeletal disorders for joint disorders, e.g. arthritis, arthrosis
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/04Centrally acting analgesics, e.g. opioids
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P29/00Non-central analgesic, antipyretic or antiinflammatory agents, e.g. antirheumatic agents; Non-steroidal antiinflammatory drugs [NSAID]
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/12Antivirals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P37/00Drugs for immunological or allergic disorders
    • A61P37/02Immunomodulators
    • A61P37/06Immunosuppressants, e.g. drugs for graft rejection
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P43/00Drugs for specific purposes, not provided for in groups A61P1/00-A61P41/00
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D413/00Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D413/14Heterocyclic compounds containing two or more hetero rings, at least one ring having nitrogen and oxygen atoms as the only ring hetero atoms containing three or more hetero rings

Definitions

  • the present invention relates to new compounds, in particular new tetrahidroisoquinolyl derivatives, to processes for their preparation and to pharmaceutical compositions containing them.
  • These compounds are potent agonists of S1 P1 receptors and thus, they are useful in the treatment, prevention or suppression of diseases and disorders known to be susceptible to improvement by sphingosine-1 -phosphate receptors agonists (S1 P1 ), such as autoimmune diseases, chronic immune and inflammatory diseases, transplant rejection, malignant neoplastic diseases, angiogenic-related disorders, pain, neurological diseases, viral and infectious diseases.
  • S1 P1 sphingosine-1 -phosphate receptors agonists
  • Sphingosine -1 phosphate is a pleiotropic lipid mediator that exhibits a broad spectrum of biological activities, including cell proliferation, survival, lymphocyte trafficking, cytoskeletal organization, and morphogenesis.
  • S1 P is generated from endogenous sphingosine through phosphorylation by specific kinases, named sphingosine kinases 1 and 2.
  • the levels of S1 P in biological fluids and tissues are tightly regulated by the balance between its synthesis by sphingosine kinases and its degradation by S1 P lyase. This tight control is important since an excessive production of S1 P has been associated to various pathological conditions, such as angiogenesis and vascular permeability changes in cancer, inflammation, myocardial infarction or transplant rejection.
  • S1 P1 to S1 P5 G-protein coupled receptor subtypes
  • S1 P1 to S1 P5 G-protein coupled receptor subtypes
  • the interest on this family of receptors increased following the discovery that they were the pharmacological target of FTY720.
  • This compound a synthetic analog of a natural product derived from the fungus lsaria sinclairii, exhibited a peculiar immunomodulatory potential in vivo. When administered in vivo, it caused lymphopenia, due to the sequestration of lymphocytes from the blood into the lymph nodes and Peyer ' s patches.
  • FTY720P phosphorylated FTY720 in vivo
  • FTY720 is currently in phase III trials for the treatment of relapsing-remitting multiple sclerosis. The drug is presumed to act by causing the retention of pathogenic lymphocytes in lymph nodes, thus preventing them to infiltrate the central nervous system (CNS).
  • CNS central nervous system
  • S1 P1 agonists have been recently disclosed for the treatment or prevention of autoimmune diseases, such as multiple sclerosis (WO2008000419, WO2008021532), rheumatoid arthritis or Crohn ' s disease (WO2007091501 ), chronic immune and inflammatory diseases such as asthma, transplant rejection (WO199400943), cancer (WO2003097028), lymphoid malignancies (WO2007143081), angiogenic-related disorders, pain (WO2004110421 , WO2007089715) neurological diseases such as neurodegeneration (WO2005025553) or dementia (WO2005058295), cardiovascular diseases (WO2004010987),.
  • autoimmune diseases such as multiple sclerosis (WO2008000419, WO2008021532), rheumatoid arthritis or Crohn ' s disease (WO2007091501 ), chronic immune and inflammatory diseases such as asthma, transplant rejection (WO199400943), cancer (WO2003097028), lymphoid malignancies (WO2007143081),
  • Autoimmune diseases include but are not limited to rheumatoid arthritis, multiple sclerosis, inflammatory bowel diseases such as Crohn's diseases and ulcerative colitis, psoriatic arthritis, thyroiditis such as Hashimoto's thyroiditis, type I diabetes; systemic lupus erythematosis and Sj ⁇ gm's syndrome.
  • Transplant rejections include but are not limited to rejections of kidney, liver, heart, lung, pancreas, cornea and skin transplants and graft-versus-host disease brought about by stem cell transplantation.
  • Immune and inflammatory diseases which may be prevented or treated include but are not limited to asthma, COPD, respiratory distress syndrome, acute or chronic pancreatitis and hepatitis; chronic sarcoidosis, contact dermatitis, atopic dermatitis, allergic rhinitis, allergic conjunctivitis, Behcet syndrome, inflammatory eye conditions such as conjunctivitis and uveitis.
  • Malignant neoplastic diseases that may be prevented or treated include but are not limited to solid cancer, tumor metastasis and lymphoid malignancies.
  • Angiogenesis-related disorders that may be prevented or treated include but are not limited to hemangiomas, ocular neovascularization, macular degeneration or diabetic retinopathy.
  • Pain, including neuropathic pain, that may be prevented or treated includes but is not limited to prophylaxis or treatment of chronic pain, wherein chronic pain is selected from chronic muscular diseases such as back pain, pain during menstruation, pain during osteoarthritis, pain during rheumatoid arthritis, pain during gastrointestinal inflammation, pain during inflammation of the heart muscle, pain during multiple sclerosis, pain during neuritis, pain during AIDS, pain during chemotherapy, tumor pain, neuropathic pain e. g. after amputation, trigeminal neuralgia, migraine or post herpetic neuralgia.
  • Cardiovascular diseases which may be prevented or treated include but are not limited to chronic heart failure, congestive heart failure, arrhythmia or tachyarrythmia, unstable angina, acute myocardial infarction and complications from cardiac surgery. Cardiovascular diseases may also refer to improving heart energy efficiency or cardiac output.
  • Neurological diseases including neurodegeneration, dementia or brain degeneration that may be prevented or treated include but are not limited to neurological disorders including Parkinson's desease, Parkinsonian disorders, Huntington's disease, Alzheimer's disease, amyotrophic lateral sclerosis, spinal ischemia, ischemic stroke, spinal cord injury, cancer-related brain injury, and cancer-related spinal cord injury, Shy-Drager syndrome, progressive supranuclear palsy, Lewy body disease, stroke, cerebral infarction, multi-infarct dementia, and geriatric dementia,
  • neurological disorders including Parkinson's desease, Parkinsonian disorders, Huntington's disease, Alzheimer's disease, amyotrophic lateral sclerosis, spinal ischemia, ischemic stroke, spinal cord injury, cancer-related brain injury, and cancer-related spinal cord injury, Shy-Drager syndrome, progressive supranuclear palsy, Lewy body disease, stroke, cerebral infarction, multi-infarct dementia, and geriatric dementia,
  • Viral diseases which may be prevented or treated include but are not limited to HIV infection, hepatitis C and cytomegalovirus infection.
  • Infectious diseases which may be prevented or treated include but are not limited to pathogenic fungal diseases.
  • tetrahidroisoquinolyls derivatives are novel and potent agonists of S1P1 and can therefore be used in the treatment or prevention of these diseases.
  • the present invention is directed to new tetrahidroisoquinolyls derivatives of formula (I) or pharmaceutically acceptable salts or N-oxides thereof
  • A is selected from the group consisting of -N-, -O- and -S-;
  • B and C independently are selected from the group consisting of -N- and -O-, with the proviso that at least two of A 1 B and C are nitrogen atoms;
  • G 1 is selected from the group consisting of nitrogen atoms and -CR C - groups, wherein R c represents a hydrogen atom, a halogen atom, a Ci -4 alkyl group or a Ci -4 alkoxy group;
  • R 1 is selected from the group consisting of hydrogen atoms, Ci -4 alkyl groups, C 1 ⁇ alkoxy groups, C 3-4 cycloalkyl groups, and -NR d R e groups wherein R d and R e are independently selected from hydrogen atoms and C 1-4 alkyl groups;
  • R 2 and R 3 are independently selected from the group consisting of hydrogen atoms and C 1-4 alkyl groups
  • R 4 , R 5 and R 7 are independently selected from the group consisting of hydrogen atoms, halogen atoms, C 1-4 alkyl groups, Ci -4 alkoxy groups and haloalkyl groups;
  • R 6 represents a C 1-4 alkyl group or a C 1-4 hydroxyalkyl group; or R 6 is selected from the group consisting of -S(O) 2 -NR a R b groups, groups, -(CH 2 ) n -NR a R b groups, -O-(CH 2 ) n -NR a R b groups, -(CH 2 J n -COOH groups, -(CH 2 ) n -NR a - CO-R b' groups, -(CH 2 ) n -NR a -(CH 2 ) p -(NH) q -SO-CH 3 groups and -(CH 2 ) n -CO-NR a R b groups, wherein
  • n, p, x and y are each independently integers from 0 to 3; q is 0 or 1 ;
  • R f , R 9 , R h , R', R J and R k independently represent hydrogen atoms or halogen atoms
  • R b is selected from the group consisting of methylsulphonyl groups, C 1-4 alkyl groups, Ci -4 hydroxyalkyl groups, C 1-4 carboxyalkyl groups, and C 1-4 haloalkyl groups;
  • R a and R b are independently selected from the group consisting of hydrogen atoms, methylsulphonyl groups, C 1-4 alkyl groups, C 1 ⁇ hydroxyalkyl groups, C 1-4 carboxyalkyl groups, and C 1-4 haloalkyl groups, or
  • R a and R b or R a and R b together with the nitrogen atom to which they are attached form a 4 to 6 membered, saturated heterocyclic group, which contains, as heteroatoms, one or two nitrogen atoms and which is substituted by a carboxyl group or a C 1-4 carboxyalkyl group; or
  • R c together with R 6 form a C 5-8 carbocyclic ring optionally substituted by -NHR' wherein R' represents a hydrogen atom or a C 1-4 carboxyalkyl group.
  • Further objectives of the present invention are to provide a method for preparing said compounds; pharmaceutical compositions comprising an effective amount of said compounds; the use of the compounds in the manufacture of a medicament for the treatment of pathological conditions or diseases susceptible to improvement by sphingosine-1 -phosphate receptors agonists (S1 P1 ), wherein the pathological condition or disease is selected from autoimmune diseases, chronic immune and inflammatory diseases, transplant rejection, malignant neoplastic diseases, angiogenic- related disorders, pain, neurological diseases.viral and infectious diseases, and methods of treatment of pathological conditions or diseases susceptible to amelioration by sphingosine-1 -phosphate receptors agonists (S1 P1 ), wherein the pathological condition or disease is selected from autoimmune diseases, chronic immune and inflammatory diseases, transplant rejection, malignant neoplastic diseases, angiogenic- related disorders, pain, neurological diseases.viral and infectious diseases comprising the administration of a therapeutically effective amount of the compounds of the invention to a subject in need of such treatment.
  • C 1-4 alkyl embraces optionally substituted, linear or branched hydrocarbon radicals having 1 to 4 carbon atoms.
  • Preferred substituents on the alkyl groups are halogen atoms and hydroxy groups, and are more preferably halogen atoms. Examples include methyl, ethyl, ⁇ -propyl, /-propyl, n-butyl, /-butyl, sec-butyl and te/f-butyl radicals.
  • C 1-4 haloalkyl group is an alkyl group, for example a C 1-4 or C 1- 2 alkyl group, which is attached to 1 , 2 or 3 halogen atoms.
  • said C 1-4 haloakyl group is chosen from -C(-)CI-, -CHCI-, -CCI 2 -, -CHCI 2 , -CCI 3 , -C(-)F-, -CHF-, -CF 2 -, -CHF 2 , -CF 3 and -CH 2 CHF 2 .
  • said C 1 ⁇ haloalkyl group is chosen from -CH 2 CI, -CHCI 2 , -CCI 3 , -CH 2 F, -CHF 2 , -CF 3 and -CH 2 CHF 2 , most preferably - CH 2 CHF 2 .
  • C 1-4 hydroxyalkyl embraces linear or branched alkyl radicals having 1 to 4 carbon atoms, any one of which may be substituted with one or more hydroxyl radicals.
  • examples of such radicals include hydroxymethyl, hydroxyethyl, hydroxypropyl, hydroxybutyl, dihydroxyethyl, 1 ,3-dihydroxypropyl, 2,3-dihydroxypropyl, 1 ,3-dihydroxy isopropyl, 2,3-dihydroxybutyl and 3,4-dihydroxybutyl radicals.
  • C 1-4 alkoxy (or alkyloxy) embraces optionally substituted, linear or branched oxy-containing radicals each having alkyl portions of 1 to 4 carbon atoms. More preferred alkoxy radicals are alkoxy radicals having 1 to 2 carbon atoms.
  • An alkoxy group is typically unsubstituted or substituted with 1 , 2 or 3 substituents which may be the same or different.
  • the substituents are preferably selected from halogen atoms, preferably chlorine or fluorine atoms and hydroxy groups. Typically, the substituents on an alkoxy group are themselves unsubstituted.
  • Preferred alkoxy radicals include methoxy, ethoxy, n-propoxy, i-propoxy, n-butoxy, sec-butoxy, t-butoxy, trifluoromethoxy, difluoromethoxy, hydroxymethoxy, 2-hydroxyethoxy and 2- hydroxypropoxy.
  • carboxyl embraces a -COOH group.
  • C 1-4 carboxyalkyl group embraces optionally substituted, linear or branched carboxyl-containing radicals having 1 to 4 carbon atoms. Preferred substituents on the alkyl group of the carboxyalkyl groups are halogen atoms and hydroxy groups.
  • cycloalkyl embraces saturated carbocyclic radicals and, unless otherwise specified, a cycloalkyl radical typically has from 3 to 6 carbon atoms, preferably from 3 to 4 carbon atoms. Examples include cyclopropyl, cyclobutyl, cyclopentyl and cyclohexyl.
  • a cycloalkyl radical When a cycloalkyl radical carries 2 or more substituents, the substituents may be the same or different.
  • Preferred substituents on the cycloalkyl groups are halogen atoms and hydroxy groups, and are more preferably halogen atoms.
  • heterocyclic group embraces typically a non-aromatic, saturated or unsaturated 4 to 6 membered ring system, in which one or more, for example 1 , 2, or 3 of the carbon atoms preferably 1 or 2 of the carbon atoms are replaced by nitrogen. Saturated heterocyclyl radicals are preferred.
  • a heterocyclic radical may be a single ring or two or more fused rings wherein at least one ring contains a heteroatom. When a heterocyclyl radical carries 2 or more substituents, the substituents may be the same or different.
  • a said optionally substituted heterocyclic group is typically unsubstituted or substituted with 1 or 2 substituents which may be the same or different.
  • the substituents are preferably a carboxyl group or a C 1-4 carboxyalkyl group.
  • the substituents on a heterocyclyl group are themselves unsubstituted.
  • the heterocyclic group carries 2 or more substituents, the substituents may be the same or different.
  • heterocyclic radicals include azetidyl, piperidyl, pyrrolidyl, pyrrolinyl, piperazinyl, pyrazolinyl and pyrazolidinyl. Azetidyl and piperazinyl are preferred.
  • atoms, radicals, moieties, chains or cycles present in the general structures of the invention are "optionally substituted".
  • substituents can be either unsubstituted or substituted in any position by one or more, for example 1 , 2, 3 or 4, substituents, whereby the hydrogen atoms bound to the unsubstituted atoms, radicals, moieties, chains or cycles are replaced by chemically acceptable atoms, radicals, moieties, chains or cycles.
  • substituents may be the same or different.
  • halogen atom embraces chlorine, fluorine, bromine or iodine atoms typically a fluorine, chlorine or bromine atom, most preferably chlorine or fluorine.
  • halo when used as a prefix has the same meaning.
  • the term pharmaceutically acceptable salt embraces salts with a pharmaceutically acceptable acid or base.
  • Pharmaceutically acceptable acids include both inorganic acids, for example hydrochloric, sulphuric, phosphoric, diphosphoric, hydrobromic, hydroiodic and nitric acid and organic acids, for example citric, fumaric, maleic, malic, mandelic, ascorbic, oxalic, succinic, tartaric, benzoic, acetic, methanesulphonic, ethanesulphonic, benzenesulphonic or p-toluenesulphonic acid.
  • Pharmaceutically acceptable bases include alkali metal (e.g. sodium or potassium) and alkali earth metal (e.g. calcium or magnesium) hydroxides and organic bases, for example alkyl amines, arylalkyl amines and heterocyclic amines.
  • X- may be an anion of various mineral acids such as, for example, chloride, bromide, iodide, sulphate, nitrate, phosphate, or an anion of an organic acid such as, for example, acetate, maleate, fumarate, citrate, oxalate, succinate, tartrate, malate, mandelate, trifluoroacetate, methanesulphonate and p-toluenesulphonate.
  • mineral acids such as, for example, chloride, bromide, iodide, sulphate, nitrate, phosphate
  • organic acid such as, for example, acetate, maleate, fumarate, citrate, oxalate, succinate, tartrate, malate, mandelate, trifluoroacetate, methanesulphonate and p-toluenesulphonate.
  • X- is preferably an anion selected from chloride, bromide, iodide, sulphate, nitrate, acetate, maleate, oxalate, succinate or trifluoroacetate. More preferably X- is chloride, bromide, trifluoroacetate or methanesulphonate.
  • an N-oxide is formed from the tertiary basic amines or imines present in the molecule, using a convenient oxidising agent.
  • R a and R b together with the nitrogen atom to which they are attached do not form a 4 to 6 membered, saturated heterocyclic group, which contains, as heteroatoms, one or two nitrogen atoms and which is substituted by a carboxyl group or a Ci -4 carboxyalkyl group
  • R a and R b are independently selected from the group consisting of hydrogen atoms, methylsulphonyl groups, Ci -4 alkyl groups, C ⁇ hydroxyalkyl groups, and C 1-4 carboxyalkyl groups
  • R a and R b each independently represents a C 1-4 alkyl group or a Ci -4 haloalkyl group.
  • A is selected from the group consisting of -N- and -O-.
  • A represents -N-.
  • the dotted line in the ring containing A, B and C denotes that the ring may be saturated, unsaturated or aromatic.
  • the ring is aromatic, i.e. is represented by the formula
  • R a and R b are not simultaneously methylsulphonyl groups.
  • R a is hydrogen and R b is selected from the group consisting of hydrogen atoms, methylsulphonyl groups, C 1-4 alkyl groups, C 1-4 hydroxyalkyl groups, C 1-4 carboxyalkyl groups, and C 1 ⁇ haloalkyl groups, or R a and R b together with the nitrogen atom to which they are attached form a 4 to 6 membered, saturated heterocyclic group, which contains, as heteroatoms, one or two nitrogen atoms and which is substitute
  • R a is hydrogen and R b is selected from the group consisting of hydrogen atoms, methylsulphonyl groups, C 1-4 alkyl groups, C 1-4 carboxyalkyl groups, and C 1-4 haloalkyl groups, or R a and R b together with the nitrogen atom to which they are attached form a 4 to 6 membered, saturated heterocyclic group, which contains, as heteroatoms, one or two nitrogen atoms and which is substituted by a carboxyl group
  • R a is hydrogen and R b is selected from the group consisting of hydrogen atoms, methylsulphonyl groups, C 1-2 carboxyalkyl groups, and C 1-2 haloalkyl groups, or R a and R b together with the nitrogen atom to which they are attached form an azetidine or piperazine group which groups are substituted by a carboxyl group or a C ⁇ 2 carboxyalkyl group.
  • G 1 represents a -CR C - group, wherein R c represents a hydrogen atom, chlorine atom, fluorine atom or a C 1 ⁇ alkyl group.
  • G 1 represents a -CR C - group, wherein R c represents a hydrogen atom or a methyl group.
  • R 1 represents a hydrogen atom, a C 1-4 alkyl group or a C 3-4 cycloalkyl group.
  • R 1 is selected from the group consisting of hydrogen atoms, ethyl groups and C 3-4 cycloalkyl groups.
  • R 2 represents a hydrogen atom or a C 1-4 alkyl group, preferably R 2 represents a hydrogen atom or a methyl group. More typically R 2 represents a C 1-4 alkyl group.
  • R 3 represents a hydrogen atom or a C 1-4 alkyl group, preferably R 3 represents a hydrogen atom or a methyl group. More typically R 3 represents a C 1-4 alkyl group.
  • R 4 represents a hydrogen atom, a C 1-4 alkyl group or a -CF 3 group, preferably a hydrogen atom or a methyl group, more preferably a hydrogen atom.
  • R 5 represents a hydrogen atom, a C 1-4 alkyl group or a -CF 3 group, preferably a hydrogen atom or a C 1-4 alkyl group, more preferably a hydrogen atom or a methyl group, being the most preferred a methyl group.
  • R 7 represents a hydrogen atom, a C 1-4 alkyl group or a -CF 3 group, preferably a hydrogen atom or a methyl group, more preferably a hydrogen atom.
  • R 4 , R 5 and R 7 is a hydrogen atom. More typically, 1 or 2 of R 4 , R 5 and R 7 are hydrogen atoms. Preferably, two of R 4 , R 5 and R 7 are hydrogen atoms. More preferably, two of R 4 , R 5 and R 7 are hydrogen atoms and the other is methyl. Even more preferably, R 4 , and R 7 are hydrogen atoms and R 5 is methyl.
  • R 6 represents a C 1 ⁇ , alkyl group, or a C 1-4 hydroxyalkyl group; or R 6 is selected from the group consisting of -(CH 2 ) n -NR a R b groups, -(CH 2 XrCOOH groups, -(CH 2 ) n -NR a -(CH 2 ) p -(NH) q -SO-CH 3 groups and -(CH 2 ) n -C0-NR a R b groups, wherein
  • n and p are each independently integers from 0 to 3
  • R a and R b are independently selected from the group consisting of hydrogen atoms, methylsulphonyl groups, C 1-4 carboxyalkyl groups, C 1-4 alkyl groups and C 1 ⁇ haloalkyl groups, or
  • R a and R b together with the nitrogen atom to which they are attached form a 4 to 6 membered, saturated heterocyclic group, which contains, as heteroatoms, one or two nitrogen atoms and which is substituted by a carboxyl group or a C 1-4 carboxyalkyl group.
  • A represents -N-
  • G 1 represents a -CR C - group, wherein R c represents a hydrogen atom or C 1-4 alkyl group;
  • R 2 and R 3 independently represent C 1-4 alkyl groups
  • R 4 , R 5 and R 7 are independently selected from the group consisting of hydrogen atoms and C 1-4 alkyl groups;
  • R 6 represents a C 1-4 alkyl group or a C 1-4 hydroxyalkyl group, or R 6 is selected from the group consisting of -S(O) 2 -NR a R b groups, -(CH 2 ) n -NR a R b groups, -(CH 2 ) n -COOH groups, -(CH 2 ) n -NR a -(CH 2 ) p -(NH) q -SO-CH 3 groups and -(CH 2 ) n -CO-NR a R b groups, wherein
  • n and p are each independently integers from 0 to 3
  • R a and R b independently are selected from the group consisting of hydrogen atoms, methylsulphonyl groups, Ci -4 carboxyalkyl groups, C 1 ⁇ , alkyl groups and Ci -4 haloalkyl groups, or
  • R a and R b together with the nitrogen atom to which they are attached form a 4 to 6 membered, saturated heterocyclic group, which contains, as heteroatoms, one or two nitrogen atoms and which is substituted by a carboxyl group or a C 1-4 carboxyalkyl group; or
  • R c together with R 6 form a C 5-8 carbocyclic ring optionally substituted by -NHR' wherein R' represents a hydrogen atom or a C 1-4 carboxyalkyl group.
  • compounds of the present invention are compounds of formula (I 1 ), or a pharmaceutically acceptable salts or N-oxides thereof,
  • G 1 is selected from the group consisting of nitrogen atoms and -CR C - groups, wherein R c represents a hydrogen atom, a chlorine atom, or a Ci -2 alkyl group;
  • R 1 is selected from the group consisting of hydrogen atoms, Ci -2 alkyl groups, cyclopropyl groups, -NH 2 groups, -NHMe, and -NMe 2 groups;
  • R 4 , R 5 and R 7 are independently selected from the group consisting of hydrogen atoms, chlorine atoms, C 1-2 alkyl groups;
  • R 6 represents a Ci. 2 alkyl group or a C 1-4 hydroxyalkyl group; or R 6 is selected from the group consisting of -S(O) 2 -NHR 6 groups, -(CH 2 ) n -NHR b groups, -(CH 2 ) n -COOH groups, -(CH 2 ) n -CO-NHR b groups, and -O-(CH 2 ) n -NHR b groups wherein n is 2 or 3,
  • R b is selected from the group consisting of hydrogen atoms, methylsulphonyl groups, C 1-2 carboxyalkyl groups, and C 1-2 haloalkyl groups, or
  • R a and R b together with the nitrogen atom to which they are attached form an azetidine or piperazine group which groups are substituted by a carboxyl group or a C 1-2 carboxyalkyl group; or
  • R c together with R 6 form a C6 carbocyclic ring substituted by -NHR' wherein R' represents a hydrogen atom or a C 1-2 carboxyalkyl group.
  • compounds of the present invention are compounds of formula (I 1 ), or pharmaceutically acceptable salts or N-oxides thereof,
  • G 1 is selected from the group consisting of nitrogen atoms and -CR C - groups, wherein R c represents a hydrogen atom, a chlorine atom, or a C 1-2 alkyl group;
  • R 1 is selected from the group consisting of hydrogen atoms, C 1-2 alkyl groups, cyclopropyl groups, and -NH 2 groups;
  • R 4 , R 5 and R 7 are independently selected from the group consisting of hydrogen atoms, chlorine atoms, Ci -2 alkyl groups;
  • R 6 represents a C 1-2 alkyl group or a C 1-4 hydroxyalkyl group; or R 6 is selected from the group consisting of -S(O) 2 -NHR" groups, -(CH 2 ) n -NHR b groups, -(CH 2 ) n -COOH groups, and -(CH 2 ) n -CO-NHR b groups, wherein n is 2 or 3,
  • R b is selected from the group consisting of hydrogen atoms, methylsulphonyl groups, C 1-2 carboxyalkyl groups, and C 1-2 haloalkyl groups, or
  • R a and R b together with the nitrogen atom to which they are attached form an azetidine or piperazine group which groups are substituted by a carboxyl group or a C 1-2 carboxyalkyl group; or
  • R c together with R 6 form a C6 carbocyclic ring substituted by -NHR' wherein R' represents a hydrogen atom or a C 1-2 carboxyalkyl group.
  • R 6 represents a C 1-4 alkyl group, or a C 1-4 hydroxyalkyl group, or
  • R 6 is selected from the group consisting of -S(O) 2 -NR a R b groups, -(CH 2 ) n -NR a R b groups, -(CH 2 XrCOOH groups, and -(CH 2 ) n -C0-NR a R b groups, wherein,
  • n is an integer from 0 to 3
  • R a and R b are independently selected from the group consisting of hydrogen atoms, methylsulphonyl groups and C 1-4 carboxyalkyl groups, C 1-4 alkyl groups and C 1-4 haloalkyl groups, or
  • R a and R b together with the nitrogen atom to which they are attached form a 4 to 6 membered, saturated heterocyclic group, which contains, as heteroatoms, one or two nitrogen atoms and which is substituted by a carboxyl group or a C 1 ⁇ , carboxyalkyl group; or
  • R c together with R 6 form a C 5-8 carbocyclic ring optionally substituted by -NHR' wherein R' represents a hydrogen atom or a C 1-4 carboxyalkyl group.
  • both A and B represent -N- and C represents -O-, G 1 represents a -CR C - group wherein R c represents a C 1-4 alkyl group,
  • R 1 represents a -NR d R e group, wherein R d and R e are independently selected from hydrogen atom and C 1-4 alkyl group, Both R 2 and R 3 represent a methyl group, Both R 4 and R 7 represent a hydrogen atom, R 5 represents a C 1-4 alkyl group, and
  • R 6 represents a C 1-4 hydroxyalkyl group, -O-(CH 2 ) n -NR a R b or -(CH 2 ) n -NR a R b , wherein R a and R b independently represent a hydrogen atom or a C 1-4 alkyl group.
  • Particular individual compounds of the invention include:
  • compounds of general formula (I) may be prepared by reacting a compound of general formula (II) wherein X can be OH or Cl, with a compound of structure (III) in a one pot reaction in solvent such as DMF, THF, etc., optionally in the presence of one or more coupling agents such as 2-(1 H-benzotriazole-1-yl)-1 ,2,3,3-tetramethyluronium tetrafluoroborate (TBTU), N-(3-dimethylaminopropyl)-N'-ethyl-carbodiimide (EDC), dicyclohexyl carbodiimide (DCC), 1-hydroxybenzotriazole (HOBt), O-(benzotriazol-1- yl)-N,N,N',N'-tetramethyluroniium hexafluorophosphate (HBTU), carbonyl diimidazole (CDI), etc.
  • solvent such as DMF, THF, etc.
  • An alternative method could be following a two steps synthesis by a first coupling as described before and a subsequent cyclization in a solvent such as xylene, toluene, benzene, pyridine, DMF, dichloromethane, acetic acid, trifluoroacetic acid, etc. at rt or elevated temperatures optionally in the presence of auxiliaries such as acid (e.g.
  • trifluoroacetic acid acetic acid, hydrochloric acid, etc.
  • bases e.g., sodium hydride, sodium acetate, sodium carbonate, potassium carbonate, triethylamine, etc.
  • tetralkylammonium salts or water removing agents such as oxalyl chloride, a carboxylic acid anhydride, phosphoryl trichloride (POCI 3 ), tetrabutylammonium fluoride (TBAF), molecular sieves, etc.
  • POCI 3 phosphoryl trichloride
  • TBAF tetrabutylammonium fluoride
  • Intermediates of formula (III) and (IV) may be obtained from the corresponding intermediates of formula (Vl) and (VII) wherein Y is CN, COOH, COCI or COOR' by reacting with hydroxylamine or hydrazine or one of its salts in a solvent such as THF, methanol, ethanol, pyridine, etc., optionally in the presence of a base such as sodium bicarbonate, sodium carbonate, potassium carbonate, triethylamine, sodium ethoxide etc. and at a temperature from room temperature to the boiling point of the solvent.
  • a solvent such as THF, methanol, ethanol, pyridine, etc.
  • a base such as sodium bicarbonate, sodium carbonate, potassium carbonate, triethylamine, sodium ethoxide etc.
  • the reaction can be carried out in the presence of a coupling agent such as 2-(1 H-benzotriazole-1-yl)-1 ,2,3,3-tetramethyluronium tetrafluoroborate, N-(3- dimethylaminopropyl)-N'-ethyl-carbodiimide, dicyclohexyl carbodiimide, 1- hydroxybenzotriazole, etc.
  • a coupling agent such as 2-(1 H-benzotriazole-1-yl)-1 ,2,3,3-tetramethyluronium tetrafluoroborate, N-(3- dimethylaminopropyl)-N'-ethyl-carbodiimide, dicyclohexyl carbodiimide, 1- hydroxybenzotriazole, etc.
  • a coupling agent such as 2-(1 H-benzotriazole-1-yl)-1 ,2,3,3-tetramethyluronium tetrafluoroborate
  • the oxadiazole compounds of general formula (Ia) may be prepared by the condensation of the tetrahidroisoquinoline-4-carboxylic acid (Ma) with the corresponding carboximidamide (Ilia) following the synthetic procedures described above.
  • the intermediates of general formula (IX) may be prepared by the reaction of the corresponding commercial ketone (VIII) with diethylcarbonate in a basic media such as sodium hydride and in an aprotic solvent such as THF or DMF and at a temperature between 0 and the boiling point of the solvent.
  • the intermediates of general formula (X) may be prepared by the condensation of the intermediates of formula (IX) with 2-cyanoacetamide in basic media such as KOH or NaOH and in a protic solvent such as methanol or ethanol at a temperature between 2O 0 C and the boiling point of the solvent.
  • the intermediates of general formula (Xl) may be prepared by the reaction of the intermediates of formula (X) with POCI 3 or a mixture of POCI 3 and PCI 5 with or without a solvent such as toluene in a high pressure reactor and at high temperature.
  • the intermediates of general formula (XII) may be obtained by the coupling of the intermediates of formula (Xl) with the corresponding alkyl boronic acid or boronate under the conditions of a Suzuki reaction (Miyaura, N.; Suzuki, A. Chem. Rev. 1995, 95, 2457).
  • This reaction may be catalyzed by a palladium catalyst like [1 ,1'- bis(diphenylphosphino)-ferrocene] dichloropalladium (II) complex with dichloromethane (1 :1 ), tetrakis(triphenylphosphine)-palladium(0), bis(triphenylphosphine)palladium(ll) chloride or tris(dibenzylideneacetone)-dipalladium(0) in an aprotic organic solvent such as dioxane, toluene, DMF or DME and in the presence of a base such as cesium carbonate, sodium carbonate, potassium carbonate or potassium phosphate at a temperature from 8O 0 C to 14O 0 C.
  • a palladium catalyst like [1 ,1'- bis(diphenylphosphino)-ferrocene] dichloropalladium (II) complex with dichloromethane (1 :1 ), t
  • the intermediates of formula (XII) can be obtained by the reaction of the intermediates of formula (Xl) with an excess of the corresponding amine or alcohol with an extra base such as NaH, triethylamine, etc. in a solvent such as MeOH, EtOH or
  • the intermediates of general formula (XVI) may be prepared by the reaction of the corresponding commercial ketone (XIV) with the corresponding acyl chloride of formula (XV) in a basic media such as lithiumdiisopropylamide (LDA) and in an aprotic solvent such as THF and at a temperature between -78 0 C and room temperature.
  • a basic media such as lithiumdiisopropylamide (LDA) and in an aprotic solvent such as THF and at a temperature between -78 0 C and room temperature.
  • the intermediates of general formula (XVII) may be prepared by the condensation of the intermediates of formula (XVI) with 2-cyanoacetamide in basic media such as triethylamine, KOH or NaOH and in a protic solvent such as methanol or ethanol at a temperature between 20 0 C and the boiling point of the solvent.
  • Intermediates of formula (Ilia) may be obtained by the reaction of hydroxylamine or one of its salts with the corresponding nitrile (XIX) in a solvent such as THF, methanol, ethanol, pyridine, etc. optionally in the presence of a base such as sodium bicarbonate, sodium carbonate, potassium carbonate, triethylamine, etc. and at a temperature from 40 to 100 0 C.
  • a solvent such as THF, methanol, ethanol, pyridine, etc.
  • a base such as sodium bicarbonate, sodium carbonate, potassium carbonate, triethylamine, etc.
  • a precursor can be the corresponding bromoaryl derivative of formula (XVIII) which react with a source of cyanide such as copper (I) cyanide, in a high boiling point solvent such as NMP, DMF or DMSO at a temperature between 150-200 0 C to yield the corresponding nitrile of general formula (XIV).
  • a source of cyanide such as copper (I) cyanide
  • a high boiling point solvent such as NMP, DMF or DMSO
  • dicyanozinc can be also used as a source of cyanide, with a Pd catalyst such as tetrakis(triphenylphosphine)-palladium(0) in a high boiling point solvent, in a standard reactor or a microwave reactor.
  • these functionalities may require temporary protection.
  • protecting groups include e.g a tert-butyl or ethyl or methyl to protect an acid, a tert- butyloxycarbonyl (BOC) to protect an amine, etc.
  • BOC tert- butyloxycarbonyl
  • protecting groups may be employed according to standard methodology (e.g. T.W. Greene, P. G. M. Wuts, Protective Groups in Organic Synthesis, 3 rd Edition, Wiley New York, 1991).
  • the compounds of general formula (Ic) wherein R 6 is a -(CH 2 ) n -CO-NR a R b group can be prepared from the corresponding acid of general formula (Ib) by reacting with ammonia, an amine or an aminoacid of general formula HNR a R b in the presence of an activating agent such as N-(3-dimethylaminopropyl)-N'-ethyl-carbodiimide (EDC), dicyclohexyl carbodiimide (DCC), 1-hydroxybenzotriazole (HOBt), (benzotriazol-1- yloxy)-tris-(dimethylamino)-phosphonium hexafluorophosphate (BOP), bis-(2-oxo-3- oxazolidinyl)-phosphonic acid chloride (BOP-CI) 1 etc. in a solvent such as THF, dioxane, DMF, dichloromethane, acetonitrile
  • primary amines of general formula (Ie) can be prepared by Curtius rearrangement of the acids of general formula (Ib) using an azide such as sodium azide, diphenylphosphoryl azide (DPPA), etc, in acidic media such as sulphuric acid or basic media such as triethylamine, in solvent such as toluene, chloroform, THF, etc. or in a protic solvent such as terf-butanol or benzyl alcohol to yield the te/t-butylcarbonyl (BOC) or benzyloxycarbonyl (CBZ or Z) protected amine and subsequent deprotection as known in the art yield the final compounds of formula (Ie).
  • an azide such as sodium azide, diphenylphosphoryl azide (DPPA), etc
  • acidic media such as sulphuric acid or basic media such as triethylamine
  • solvent such as toluene, chloroform, THF, etc.
  • the compounds of general formula (Ig) may be prepared by the reductive amination of the aldehyde derivatives of general formula (XXIV) with the corresponding amine or aminoacid of formula HNR a R b in acidic media such as acetic acid, in a protic solvent such as methanol or ethanol and with a reductive agent such as sodium borohydride or sodium cyanoborohydride at a temperature from O 0 C to the boiling point of the solvent.
  • Intermediates of formula (XXIV) may be obtained by oxidation of diols of general formula (If) with an oxidative reagent such as NaIO 4 , NaCIO 4 , KIO 4 , etc. in a solvent such as methanol, ethanol, THF, dioxane, ether, etc. with the presence or absence of water.
  • an oxidative reagent such as NaIO 4 , NaCIO 4 , KIO 4 , etc. in a solvent such as methanol, ethanol, THF, dioxane, ether, etc. with the presence or absence of water.
  • Compounds of general formula (If) may be prepared by oxidation of the allyl intermediates of general formula (XXIII) using an oxidative reagent such as N-methyl- morpholine-N-oxide, sodium periodate, sodium perchlorate, potassium periodate, etc. in the presence of a catalyst such us sodium tetraoxide solid or a solution in water or tert-butanol, in a solvent such as methanol, ethanol, THF, dioxane, ether, acetone, etc. with the presence or absence of water.
  • an oxidative reagent such as N-methyl- morpholine-N-oxide, sodium periodate, sodium perchlorate, potassium periodate, etc.
  • a catalyst such us sodium tetraoxide solid or a solution in water or tert-butanol, in a solvent such as methanol, ethanol, THF, dioxane, ether, acetone, etc. with the presence or absence of water.
  • Allyl intermediates of general formula (XXIII) may be prepared by the condensation of the corresponding carboximidamide of formula (IHc) with the corresponding acid of general formula (Ma) as described above.
  • a catalyst such as tetrakis(triphenylphosphine)-palladium(0), palladium acetate, bis(triphenylphosphine)palladium(ll) chloride or tris(dibenzylideneacetone)- dipalladium(O)
  • the compounds of formula (Ih) may be obtained by the reaction of compounds of general formula (XXVII) with the corresponding alkylating agent in basic media such as sodium hydride in a solvent such as THF or DMF at a temperature from 0 to 15O 0 C.
  • basic media such as sodium hydride in a solvent such as THF or DMF
  • the phenolic functionality of (XXVII) may be coupled to suitable alcohol derivatives using a Mitsunobu coupling procedure (Mitsunobu, O., Synthesis 1 (1981 )).
  • Preferred coupling conditions include the use of a thalkylphosphine or triarylphosphine, such as tri-n-butylphosphine or triphenylphosphine, in a suitable solvent, such as tetrahydrofuran or dichloromethane, and an azodicarbonyl reagent, such as diethyl azodicarboxylate or 1 ,1'-(azodicarbonyl)dipipehdine.
  • a suitable solvent such as tetrahydrofuran or dichloromethane
  • an azodicarbonyl reagent such as diethyl azodicarboxylate or 1 ,1'-(azodicarbonyl)dipipehdine.
  • the compounds of general formula (XXVII) may be prepared by demethylation of the corresponding compound of general formula (XXVI) using BBr 3 or AIBr 3 or BF 3 or iodotrimethylsilane as demethylating agent in a solvent such as dichloromethane or 1 ,2-dichloroethane, chloroform at a temperature between 0 and the 6O 0 C.
  • a solvent such as dichloromethane or 1 ,2-dichloroethane, chloroform at a temperature between 0 and the 6O 0 C.
  • compounds of general formula (XXVII) may be prepared by demethylation using HBr in acetic acid as a solvent.
  • the mobile phase was formic acid (0.4 ml_), ammonia (0.1 ml_), methanol (500 tnL) and acetonitrile (500 ml.) (B) and formic acid (0.46 ml_), ammonia (0.115 ml.) and water (1000 ml.) (A), the gradients are specified in the following table for each methode used.
  • the reequilibration time between two injections was 1 min.
  • the flow rate was 0.8 mL ⁇ nin for method A and 0.4 mL/min for method B and C.
  • the injection volume was 5 microliter for method A and B and 3 microliter for method C. Diode array chromatograms were collected at 210 nM.
  • the solid was dissolved in DMSO/MeOH, injected into a Biotage C18 silica column (4OM, 25M or 25S according to the crude amount) and eluted on the SP1® automated purification system from Biotage.
  • the gradient used was H2O/Acetonitrile/MeOH (1 :1 ) (0.1% v/v HCOONH4 both phases) from 0% to 100% acetonitrile/MeOH (1 :1 ) in 80 column volumes.
  • the appropriate fractions were collected and the organic solvent evaporated under reduced pressure or liofilized.
  • Preparation 20 (1.1g, 4.43mmol) was dissolved in methanol (35ml_) and sodium acetate (0.55g, 6.70mmol) was added. Finally palladium chloride (0.16g, 0.90mmol) was added and the mixture hydrogenated at 15 psi for 5h. The catalyst was filtered off and the filtrate concentrated. The residue was redissolved in DCM and washed with water. The organic layer was dried over magnesium sulphate and concentrated to yield an oil (82% yield) as the desired compound.
  • Example 9 A mixture of Example 9 (100mg, 0.25mmol), tert-butyl 2-aminoacetate hydrochloride (62mg, 0.37mmol), HATU (94mg, 0.25mmol) and DIEA (130 ⁇ l, 0.75mmol) in DMF (3mL) was stirred overnight at r.t. The mixture was poured over ice/water and stirred for 15min. The solid formed was filtered off, washed with water and dried in the vacuum oven and used without further purification.
  • 6-bromo-1 ,2,3,4-tetrahydronaphthalen-2-amine To a solution of the 6-bromo 2-tetralone (2g, 8.89mmol) and NH 4 OAc (5.52g,
  • Example 18 A mixture of Example 18 (172mg, 0.46mmol), ethyl 2-oxoacetate (146Dl, 0.74mmol) and AcOH (239 ⁇ l, 4.18mmol) in methanol (5ml_) was stirred at room temperature for 1 h. Then NaBH 3 CN (20mg, 0.32mmol) was added and the reaction mixture stirred at r.t. overnight. Solvent was concentrated and the residue dissolved in ethyl acetate, washed with water and brine, dried over magnesium sulphate, filtered and concentrated. The crude obtained was purified according the General purification method to give the desired compound (50% yield).
  • Example 21 To a solution of Example 21 (75mg, 0.19mmol) in DCM (1OmL) was added triethylamine (94 ⁇ L 3.51 mmol) and the mixture stirred for 5 min. Then methanesulfonyl chloride (17 ⁇ L, 0.22mol) was added and the reaction mixture stirred at room temperature for 6h. Ethyl acetate was added and washed twice with water and once with brine. This organic layer was dried over magnesium sulphate and concentrated to give a white solid which was washed with diisopropyl ether and dried in the vacuum oven (66% yield).
  • the effect of the compounds was measured using a 35S-GTPyS binding assay. Briefly, membranes were incubated in a buffer containing 2OmM HEPES pH 7.4, 10OmM NaCI, 1OmM MgCI2, 10 ⁇ M GDP, 50 ⁇ g/mL saponin and 0.2% fatty acid-free BSA at various concentrations (0.1 nM-10 ⁇ M) and 0.1 nM 35S-GTPyS. 10 ⁇ M S1 P was used as 100% maximum efficacy. The assay was incubated for 90 min at room temperature with gentle mixing, and terminated by filtrating the reaction mixture through GF/C filter plates using the Manifold Filtration System. The filters were immediately washed with sodium phosphate pH 7.4 buffer. After drying the filter plates scintillant liquid were added to each well and 35S-GTPyS binding was measured on a Trilux Scintillation Counter.
  • the tetrahidroisoquinolyl derivatives of the invention may also be combined with other active compounds in the treatment of diseases known to be susceptible to improvement by treatment with a sphingosine-1 -phosphate receptor agonist (S1 P1 ).
  • S1 P1 sphingosine-1 -phosphate receptor agonist
  • the combinations of the invention can optionally comprise one or more additional active substances which are known to be useful in the treatment of autoimmune diseases, chronic immune and inflammatory diseases, transplant rejection, malignant neoplastic diseases, angiogenic-related disorders, pain, neurological diseases.
  • viral and infectious diseases such as (a) beta interferons such as Betaseron, Avonex or Rebif, (b), immunomodulators such as glatiramer acetate, (c) inhibitors of DNA synthesis and repair, such as Mitoxantrone, (d) anti-alpha 4 integrin antibodies, such as Natalizumab (Tysabri), (e) alpha 4 integrin antagonists such as R-1295 , TBC-4746, CDP-323, ELND-002, Firategrast and TMC-2003, (f), dyhydrofolate reductase inhibitors, such as Methotrexate or CH-1504, (g) glucocorticoids such as prednisone or methylpredn
  • the combinations of the invention may be used in the treatment of disorders which are susceptible to amelioration by sphingosine-1 -phosphate receptors agonists (S1 P1 ).
  • S1 P1 sphingosine-1 -phosphate receptors agonists
  • Preferred examples of such disorders are multiple sclerosis, transplant rejection, systemic lupus erythematosus, asthma, psoriasis, rheumatoid arthritis, psoriatic arthritis and Crohn's disease, more preferably multiple sclerosis, transplant rejection, asthma and rheumatoid arthritis, and most preferably multiple sclerosis.
  • the active compounds in the combinations of the invention may be administered by any suitable route, depending on the nature of the disorder to be treated, e.g. orally (as syrups, tablets, capsules, lozenges, controlled-release preparations, fast-dissolving preparations, etc); topically (as creams, ointments, lotions, nasal sprays or aerosols, etc); by injection (subcutaneous, intradermic, intramuscular, intravenous, etc.) or by inhalation (as a dry powder, a solution, a dispersion, etc).
  • suitable route e.g. orally (as syrups, tablets, capsules, lozenges, controlled-release preparations, fast-dissolving preparations, etc); topically (as creams, ointments, lotions, nasal sprays or aerosols, etc); by injection (subcutaneous, intradermic, intramuscular, intravenous, etc.) or by inhalation (as a dry powder, a solution, a dispersion,
  • the active compounds in the combination i.e. the sphingosine-1 -phosphate agonist of the invention, and the other optional active compounds may be administered together in the same pharmaceutical composition or in different compositions intended for separate, simultaneous, concomitant or sequential administration by the same or a different route.
  • One execution of the present invention consists of a kit of parts comprising a sphingosine-1 -phosphate agonist of the invention together with instructions for simultaneous, concurrent, separate or sequential use in combination with another active compound useful in the treatment of multiple sclerosis, transplant rejection, systemic lupus erythematosus, asthma, psoriasis, rheumatoid arthritis, psoriatic arthritis and Crohn's disease,
  • Another execution of the present invention consists of a package comprising a sphingosine-1 -phosphate agonist of formula (I) and another active compound useful in the treatment of multiple sclerosis, transplant rejection, systemic lupus erythematosus, asthma, psoriasis, rheumatoid arthritis, psoriatic arthritis and Crohn's disease,
  • compositions may conveniently be presented in unit dosage form and may be prepared by any of the methods well known in the art of pharmacy.
  • Formulations of the present invention suitable for oral administration may be presented as discrete units such as capsules, cachets or tablets each containing a predetermined amount of the active ingredient; as a powder or granules; as a solution or a suspension in an aqueous liquid or a non-aqueous liquid; or as an oil- in-water liquid emulsion or a water-in-oil liquid emulsion.
  • the active ingredient may also be presented as a bolus, electuary or paste.
  • a syrup formulation will generally consist of a suspension or solution of the compound or salt in a liquid carrier for example, ethanol, peanut oil, olive oil, glycerine or water with flavouring or colouring agent.
  • a liquid carrier for example, ethanol, peanut oil, olive oil, glycerine or water with flavouring or colouring agent.
  • composition is in the form of a tablet
  • any pharmaceutical carrier routinely used for preparing solid formulations may be used.
  • examples of such carriers include magnesium stearate, talc, gelatine, acacia, stearic acid, starch, lactose and sucrose.
  • a tablet may be made by compression or moulding, optionally with one or more accessory ingredients.
  • Compressed tablets may be prepared by compressing in a suitable machine the active ingredient in a free-flowing form such as a powder or granules, optionally mixed with a binder, lubricant, inert diluent, lubricating, surface active or dispersing agent.
  • Moulded tablets may be made by moulding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent.
  • the tablets may optionally be coated or scored and may be formulated so as to provide slow or controlled release of the active ingredient therein.
  • composition is in the form of a capsule
  • any routine encapsulation is suitable, for example using the aforementioned carriers in a hard gelatine capsule.
  • composition is in the form of a soft gelatine capsule
  • any pharmaceutical carrier routinely used for preparing dispersions or suspensions may be considered, for example aqueous gums, celluloses, silicates or oils, and are incorporated in a soft gelatine capsule.
  • Dry powder compositions for topical delivery to the lung by inhalation may, for example, be presented in capsules and cartridges of for example gelatine or blisters of for example laminated aluminium foil, for use in an inhaler or insufflator.
  • Formulations generally contain a powder mix for inhalation of the compound of the invention and a suitable powder base (carrier substance) such as lactose or starch. Use of lactose is preferred.
  • a suitable powder base such as lactose or starch.
  • lactose is preferred.
  • Each capsule or cartridge may generally contain between 2 ⁇ g and 150 ⁇ g of each therapeutically active ingredient.
  • the active ingredient (s) may be presented without excipients.
  • Packaging of the formulation for inhalation may be carried out by using suitable inhaler devices such as the NOVOLIZER ® SD2FL or GENUAIR ® which are described in the following patent applications: WO 97/000703, WO 03/000325 and WO 03/061742.
  • suitable inhaler devices such as the NOVOLIZER ® SD2FL or GENUAIR ® which are described in the following patent applications: WO 97/000703, WO 03/000325 and WO 03/061742.
  • compositions for nasal delivery include those mentioned above for inhalation and further include non-pressurized compositions in the form of a solution or suspension in an inert vehicle such as water optionally in combination with conventional excipients such as buffers, anti-microbials, tonicity modifying agents and viscosity modifying agents which may be administered by nasal pump.
  • an inert vehicle such as water
  • excipients such as buffers, anti-microbials, tonicity modifying agents and viscosity modifying agents which may be administered by nasal pump.
  • Typical dermal and transdermal formulations comprise a conventional aqueous or nonaqueous vehicle, for example a cream, ointment, lotion or paste or are in the form of a medicated plaster, patch or membrane.
  • the composition is in unit dosage form, for example a tablet, capsule or metered aerosol dose, so that the patient may administer a single dose.
  • the amount of each active which is required to achieve a therapeutic effect will, of course, vary with the particular active, the route of administration, the subject under treatment, and the particular disorder or disease being treated.
  • Effective doses are normally in the range of 2-2000 mg of active ingredient per day.
  • Daily dosage may be administered in one or more treatments, preferably from 1 to 4 treatments, per day.
  • the active ingredients are administered once or twice a day.
  • active agents When combinations of actives are used, it is contemplated that all active agents would be administered at the same time, or very close in time. Alternatively, one or two actives could be taken in the morning and the other (s) later in the day. Or in another scenario, one or two actives could be taken twice daily and the other (s) once daily, either at the same time as one of the twice-a-day dosing occurred, or separately. Preferably at least two, and more preferably all, of the actives would be taken together at the same time. Preferably, at least two, and more preferably all actives would be administered as an admixture.
  • the above ingredients were sieved through a 60 mesh sieve, and were loaded into a suitable mixer and filled into 50,000 gelatine capsules.
  • COMPOSITION EXAMPLE 2 50,000 tablets, each containing 50 mg of 3-(4-(5-(1-ethyl-7,7-dimethyl-5,6,7,8- tetrahydroisoquinolin-4-yl)-1 ,2,4-oxadiazol-3-yl)-2,6-dimethylphenyl)propanamide (active ingredient), were prepared from the following formulation:
  • All the powders were passed through a screen with an aperture of 0.6 mm, then mixed in a suitable mixer for 20 minutes and compressed into 300 mg tablets using 9 mm disc and flat bevelled punches.
  • the disintegration time of the tablets was about 3 minutes.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • General Chemical & Material Sciences (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Engineering & Computer Science (AREA)
  • Immunology (AREA)
  • Communicable Diseases (AREA)
  • Oncology (AREA)
  • Neurosurgery (AREA)
  • Pulmonology (AREA)
  • Pain & Pain Management (AREA)
  • Rheumatology (AREA)
  • Biomedical Technology (AREA)
  • Neurology (AREA)
  • Orthopedic Medicine & Surgery (AREA)
  • Transplantation (AREA)
  • Physical Education & Sports Medicine (AREA)
  • Dermatology (AREA)
  • Virology (AREA)
  • Epidemiology (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
  • Plural Heterocyclic Compounds (AREA)
  • Heterocyclic Carbon Compounds Containing A Hetero Ring Having Nitrogen And Oxygen As The Only Ring Hetero Atoms (AREA)
EP10700383A 2009-01-19 2010-01-14 Oxadiazolderivate als slpl-rezeptoragonisten Withdrawn EP2387571A1 (de)

Priority Applications (1)

Application Number Priority Date Filing Date Title
EP10700383A EP2387571A1 (de) 2009-01-19 2010-01-14 Oxadiazolderivate als slpl-rezeptoragonisten

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
EP09382004A EP2210890A1 (de) 2009-01-19 2009-01-19 Oxadiazol Derivate als S1P1 Rezeptor Agonisten
PCT/EP2010/000158 WO2010081692A1 (en) 2009-01-19 2010-01-14 Oxadiazole derivatives as slpl receptor agonists
EP10700383A EP2387571A1 (de) 2009-01-19 2010-01-14 Oxadiazolderivate als slpl-rezeptoragonisten

Publications (1)

Publication Number Publication Date
EP2387571A1 true EP2387571A1 (de) 2011-11-23

Family

ID=40568602

Family Applications (2)

Application Number Title Priority Date Filing Date
EP09382004A Withdrawn EP2210890A1 (de) 2009-01-19 2009-01-19 Oxadiazol Derivate als S1P1 Rezeptor Agonisten
EP10700383A Withdrawn EP2387571A1 (de) 2009-01-19 2010-01-14 Oxadiazolderivate als slpl-rezeptoragonisten

Family Applications Before (1)

Application Number Title Priority Date Filing Date
EP09382004A Withdrawn EP2210890A1 (de) 2009-01-19 2009-01-19 Oxadiazol Derivate als S1P1 Rezeptor Agonisten

Country Status (17)

Country Link
US (1) US20110274657A1 (de)
EP (2) EP2210890A1 (de)
JP (1) JP2012515182A (de)
KR (1) KR20110110198A (de)
CN (1) CN102282144A (de)
AR (1) AR075022A1 (de)
AU (1) AU2010205825A1 (de)
BR (1) BRPI1005153A2 (de)
CA (1) CA2748394A1 (de)
EA (1) EA201101089A1 (de)
EC (1) ECSP11011200A (de)
IL (1) IL213630A0 (de)
MX (1) MX2011007455A (de)
SG (1) SG172452A1 (de)
TW (1) TW201028407A (de)
UY (1) UY32364A (de)
WO (1) WO2010081692A1 (de)

Families Citing this family (41)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
HUE030235T2 (en) 2005-12-13 2017-04-28 Incyte Holdings Corp Heteroaryl-substituted pyrrolo [2,3-b] pyridines and pyrrolo [2,3-b] pyrimidines as Janus kinase inhibitors
EP3070090B1 (de) 2007-06-13 2018-12-12 Incyte Holdings Corporation Verwendung der salze des janus-kinasehemmers (r)-3-(4-(7h-pyrrolo[2,3-d]pyrimidin-4-yl)-1h-pyrazol-1-yl)-3-cyclopentylpropannitril
AR076794A1 (es) 2009-05-22 2011-07-06 Incyte Corp Derivados de n-(hetero)aril-pirrolidina de pirazol-4-il-pirrolo [2,3-d]pirimidinas y pirrol-3-il-pirrolo [2,3-d ]pirimidinas como inhibidores de la quinasa janus y composiciones farmaceuticas que los contienen
EA020494B1 (ru) 2009-05-22 2014-11-28 Инсайт Корпорейшн 3-[4-(7H-ПИРРОЛО[2,3-d]ПИРИМИДИН-4-ИЛ)-1H-ПИРАЗОЛ-1-ИЛ]ОКТАН- ИЛИ ГЕПТАННИТРИЛ КАК JAK-ИНГИБИТОРЫ
US8399451B2 (en) 2009-08-07 2013-03-19 Bristol-Myers Squibb Company Heterocyclic compounds
AR078012A1 (es) 2009-09-01 2011-10-05 Incyte Corp Derivados heterociclicos de las pirazol-4-il- pirrolo (2,3-d) pirimidinas como inhibidores de la quinasa janus
MX347851B (es) 2010-03-10 2017-05-16 Incyte Corp Derivados de piperidin-4-il azetidina como inhibidores de janus cinasa 1 (jak1).
EP3087972A1 (de) 2010-05-21 2016-11-02 Incyte Holdings Corporation Topische formulierung für einen jak-hemmer
US8629282B2 (en) 2010-11-03 2014-01-14 Bristol-Myers Squibb Company Heterocyclic compounds as S1P1 agonists for the treatment of autoimmune and vascular diseases
EP2640725B1 (de) 2010-11-19 2015-01-07 Incyte Corporation Heterocyclensubstituierte pyrrolopyridine und pyrrolopyrimidine als jak-inhibitoren
SG190839A1 (en) 2010-11-19 2013-07-31 Incyte Corp Cyclobutyl substituted pyrrolopyridine and pyrrolopyrimidine derivatives as jak inhibitors
EP2455080A1 (de) * 2010-11-23 2012-05-23 Almirall, S.A. S1P1-Rezeptoragonisten zur Verwendung bei der Behandlung von multipler Sklerose
EP2455081A1 (de) * 2010-11-23 2012-05-23 Almirall, S.A. S1P1-Rezeptoragonisten zur Verwendung bei der Behandlung von Morbus Crohn
KR102024948B1 (ko) 2011-02-18 2019-11-04 노파르티스 파르마 아게 mTOR/JAK 저해제 병용 요법
AR086983A1 (es) 2011-06-20 2014-02-05 Incyte Corp Derivados de azetidinil fenil, piridil o pirazinil carboxamida como inhibidores de jak
WO2013023119A1 (en) 2011-08-10 2013-02-14 Novartis Pharma Ag JAK P13K/mTOR COMBINATION THERAPY
TW201313721A (zh) 2011-08-18 2013-04-01 Incyte Corp 作為jak抑制劑之環己基氮雜環丁烷衍生物
UA111854C2 (uk) 2011-09-07 2016-06-24 Інсайт Холдінгс Корпорейшн Способи і проміжні сполуки для отримання інгібіторів jak
WO2013113716A1 (en) 2012-02-03 2013-08-08 Basf Se Fungicidal pyrimidine compounds
WO2013113776A1 (en) 2012-02-03 2013-08-08 Basf Se Fungicidal pyrimidine compounds
WO2013113720A1 (en) 2012-02-03 2013-08-08 Basf Se Fungicidal pyrimidine compounds
CN104220427A (zh) 2012-02-03 2014-12-17 巴斯夫欧洲公司 杀真菌嘧啶化合物
EA201400858A1 (ru) 2012-02-03 2015-01-30 Басф Се Фунгицидные пиримидиновые соединения
WO2013113791A1 (en) 2012-02-03 2013-08-08 Basf Se Fungicidal pyrimidine compounds
WO2013113773A1 (en) 2012-02-03 2013-08-08 Basf Se Fungicidal pyrimidine compounds
AR091079A1 (es) 2012-05-18 2014-12-30 Incyte Corp Derivados de pirrolopirimidina y pirrolopiridina sustituida con piperidinilciclobutilo como inhibidores de jak
KR20220162825A (ko) 2012-11-15 2022-12-08 인사이트 홀딩스 코포레이션 룩솔리티니브의 서방성 제형
UY35338A (es) 2013-02-21 2014-08-29 Bristol Myers Squibb Company Una Corporación Del Estado De Delaware Compuestos bicíclicos moduladores de la actividad de s1p1 y composiciones farmacéuticas que los contienen
AU2014225938B2 (en) 2013-03-06 2018-07-19 Incyte Holdings Corporation Processes and intermediates for making a JAK inhibitor
KR20160045081A (ko) 2013-08-07 2016-04-26 인사이트 코포레이션 Jak1 억제제용 지속 방출 복용 형태
WO2015184305A1 (en) 2014-05-30 2015-12-03 Incyte Corporation TREATMENT OF CHRONIC NEUTROPHILIC LEUKEMIA (CNL) AND ATYPICAL CHRONIC MYELOID LEUKEMIA (aCML) BY INHIBITORS OF JAK1
UY36274A (es) 2014-08-20 2016-02-29 Bristol Myers Squibb Company Una Corporación Del Estado De Delaware Compuestos bicíclicos sustituidos como agonistas selectivos de la actividad del receptor s1p1 acoplado a la proteína g
MA41139A (fr) 2014-12-11 2017-10-17 Actelion Pharmaceuticals Ltd Combinaison pharmaceutique comportant un agoniste sélectif du récepteur sip1
US11192886B2 (en) * 2016-07-22 2021-12-07 Medshine Discovery Inc. S1P1 agonist and application thereof
TW201924683A (zh) 2017-12-08 2019-07-01 美商英塞特公司 用於治療骨髓增生性贅瘤的低劑量組合療法
TWI797242B (zh) 2018-01-30 2023-04-01 美商英塞特公司 製備jak抑制劑之方法及中間物
MX2022012285A (es) 2018-03-30 2023-08-15 Incyte Corp Tratamiento de la hidradenitis supurativa mediante el uso de inhibidores de actividad de la cinasa janus (jak).
WO2021101854A1 (en) 2019-11-19 2021-05-27 Trevena, Inc. Compounds and methods of preparing compounds s1p1 modulators
US11833155B2 (en) 2020-06-03 2023-12-05 Incyte Corporation Combination therapy for treatment of myeloproliferative neoplasms
JP2024506387A (ja) * 2021-02-10 2024-02-13 アイカーン スクール オブ メディスン アット マウント サイナイ 腎線維症を治療するためのオキサジアゾリルジヒドロピラノ[2,3‐b]ピリジン系HIPK2阻害剤
WO2024031041A2 (en) * 2022-08-05 2024-02-08 Icahn School Of Medicine At Mount Sinai Oxygen-containing heterocyclic compound and use thereof

Family Cites Families (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO1994000943A1 (en) 1992-06-23 1994-01-06 Motorola, Inc. Multi-modulation scheme compatible radio
DE69617431T2 (de) 1995-06-21 2002-08-01 Asta Medica Ag Arzneipulverkartusche mit integrierter dosiereinrichtung, sowie pulverinhalator
DE10129703A1 (de) 2001-06-22 2003-01-02 Sofotec Gmbh & Co Kg Zerstäubungssystem für eine Pulvermischung und Verfahren für Trockenpulverinhalatoren
DE10202940A1 (de) 2002-01-24 2003-07-31 Sofotec Gmbh & Co Kg Patrone für einen Pulverinhalator
CA2483594C (en) 2002-05-16 2011-02-15 Novartis Ag Use of edg receptor binding agents in cancer
GB0217152D0 (en) 2002-07-24 2002-09-04 Novartis Ag Organic compounds
WO2004103279A2 (en) * 2003-05-15 2004-12-02 Merck & Co., Inc. 3-(2-amino-1-azacyclyl)-5-aryl-1,2,4-oxadiazoles as s1p receptor agonists
EP1484057A1 (de) 2003-06-06 2004-12-08 Aventis Pharma Deutschland GmbH Verwendung von 2-amino-1,3-propandiolderivaten zur herstrllung eines medikaments zur behandlung von bestimmten schmerzen
AU2004271804B2 (en) 2003-09-12 2011-01-06 Newron Sweden Ab Treatment of disorders of the nervous system
US20070043014A1 (en) * 2003-10-01 2007-02-22 Merck & Co., Inc. 3,5-Aryl, heteroaryl or cycloalkyl substituted-1,2,4-oxadiazoles as s1p receptor agonists
WO2005058848A1 (en) * 2003-12-17 2005-06-30 Merck & Co., Inc. (3,4-disubstituted)propanoic carboxylates as s1p (edg) receptor agonists
GB0329498D0 (en) 2003-12-19 2004-01-28 Novartis Ag Organic compounds
US20080306124A1 (en) * 2005-06-08 2008-12-11 Rainer Albert Polycyclic Oxadiazoles or I Soxazoles and Their Use as Sip Receptor Ligands
GB0601744D0 (en) * 2006-01-27 2006-03-08 Novartis Ag Organic compounds
AU2007209961A1 (en) 2006-01-27 2007-08-09 University Of Virginia Patent Foundation Method for treatment of neuropathic pain
TWI389683B (zh) 2006-02-06 2013-03-21 Kyorin Seiyaku Kk A therapeutic agent for an inflammatory bowel disease or an inflammatory bowel disease treatment using a 2-amino-1,3-propanediol derivative as an active ingredient
US8318812B2 (en) 2006-06-02 2012-11-27 The Ohio State University Research Foundation Therapeutic agents for the treatment of lymphoid malignancies
GB0612721D0 (en) 2006-06-27 2006-08-09 Novartis Ag Organic compounds
CN101553216B (zh) 2006-08-17 2012-03-21 芝加哥大学 炎性疾病的治疗

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See references of WO2010081692A1 *

Also Published As

Publication number Publication date
WO2010081692A1 (en) 2010-07-22
AR075022A1 (es) 2011-03-02
EP2210890A1 (de) 2010-07-28
EA201101089A1 (ru) 2012-02-28
MX2011007455A (es) 2011-08-03
TW201028407A (en) 2010-08-01
JP2012515182A (ja) 2012-07-05
CA2748394A1 (en) 2010-07-22
AU2010205825A1 (en) 2011-07-07
IL213630A0 (en) 2011-07-31
UY32364A (es) 2010-02-26
US20110274657A1 (en) 2011-11-10
KR20110110198A (ko) 2011-10-06
SG172452A1 (en) 2011-07-28
ECSP11011200A (es) 2011-08-31
CN102282144A (zh) 2011-12-14
BRPI1005153A2 (pt) 2019-09-24

Similar Documents

Publication Publication Date Title
WO2010081692A1 (en) Oxadiazole derivatives as slpl receptor agonists
KR101721716B1 (ko) 선택적인 스핑고신 1 포스페이트 수용체 조절자 및 카이랄 합성 방법
WO2011144338A1 (en) Pyrazole derivatives as s1p1 agonists
TWI410421B (zh) 吡啶-4-基衍生物
JP2012513958A (ja) 1,2,4−オキサジアゾール誘導体およびそれらの治療的使用
KR101781233B1 (ko) 스핑고신 1 포스페이트 수용체 조절자 및 카이랄 합성 방법
CA2524867A1 (en) 3-(2-amino-1-azacyclyl)-5-aryl-1,2,4-oxadiazoles as s1p receptor agonists
TW201113277A (en) New thiadiazole derivatives
ES2548258T3 (es) Compuestos de oxadiazol sustituidos y su uso como agonistas de S1P1
CA2818470C (en) 2-methoxy-pyridin-4-yl derivatives
TW201120019A (en) New 2-aminothiadiazole derivatives
WO2011113578A1 (en) New oxadiazole derivatives
US20220194908A1 (en) Sphingosine 1 phosphate receptor modulators

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20110815

AK Designated contracting states

Kind code of ref document: A1

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HR HU IE IS IT LI LT LU LV MC MK MT NL NO PL PT RO SE SI SK SM TR

AX Request for extension of the european patent

Extension state: AL BA RS

17Q First examination report despatched

Effective date: 20120525

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20120801